WO2014044768A1 - Treatment for rheumatoid arthritis - Google Patents

Treatment for rheumatoid arthritis Download PDF

Info

Publication number
WO2014044768A1
WO2014044768A1 PCT/EP2013/069501 EP2013069501W WO2014044768A1 WO 2014044768 A1 WO2014044768 A1 WO 2014044768A1 EP 2013069501 W EP2013069501 W EP 2013069501W WO 2014044768 A1 WO2014044768 A1 WO 2014044768A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
administered
dose
csf
patient
Prior art date
Application number
PCT/EP2013/069501
Other languages
English (en)
French (fr)
Inventor
Stefan HAERTLE
Stephane Leclair
Amgad SHEBL
Stefan Steidl
Original Assignee
Morphosys Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2013320261A priority Critical patent/AU2013320261A1/en
Priority to CA2884124A priority patent/CA2884124A1/en
Application filed by Morphosys Ag filed Critical Morphosys Ag
Priority to KR1020217005022A priority patent/KR20210021153A/ko
Priority to JP2015532412A priority patent/JP2015533806A/ja
Priority to KR1020157009911A priority patent/KR20150056846A/ko
Priority to MA37946A priority patent/MA37946B1/fr
Priority to KR1020237008704A priority patent/KR20230041086A/ko
Priority to IN657KON2015 priority patent/IN2015KN00657A/en
Priority to KR1020227006147A priority patent/KR20220028177A/ko
Priority to CN201380049042.8A priority patent/CN104995210A/zh
Priority to UAA201502131A priority patent/UA117228C2/uk
Priority to EP21169696.8A priority patent/EP3916013A1/en
Priority to EA201590359A priority patent/EA031489B1/ru
Priority to MX2015003644A priority patent/MX2015003644A/es
Priority to BR112015006189-3A priority patent/BR112015006189B1/pt
Priority to EP18155371.0A priority patent/EP3345923A1/en
Priority to SG11201501595YA priority patent/SG11201501595YA/en
Priority to EP13763100.8A priority patent/EP2897977A1/en
Priority to US14/429,996 priority patent/US20150246969A1/en
Publication of WO2014044768A1 publication Critical patent/WO2014044768A1/en
Priority to IL237554A priority patent/IL237554B/en
Priority to ZA2015/01559A priority patent/ZA201501559B/en
Priority to PH12015500591A priority patent/PH12015500591B1/en
Priority to CR20150153A priority patent/CR20150153A/es
Priority to HK15108769.5A priority patent/HK1208231A1/xx
Priority to AU2016250388A priority patent/AU2016250388B2/en
Priority to US15/880,726 priority patent/US10913792B2/en
Priority to US17/144,685 priority patent/US20210130451A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/243Colony Stimulating Factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention provides anti-GM-CSF antibodies for use in the treatment of rheumatoid arthritis, and methods for the treatment of rheumatoid arthritis using such antibodies.
  • Anti-GM-CSF antibodies, in particular MOR103, are administered to patients suffering from rheumatoid arthritis at dosages that are beneficial in a clinical setting.
  • RA Rheumatoid arthritis
  • RA primarily affects the joints and is characterized by chronic inflammation of the synovial tissue, which eventually leads to the destruction of cartilage, bone and ligaments and can cause joint deformity.
  • RA has a peak incidence between 40 and 60 years of age and affects primarily women.
  • the cause of RA is not known; however, certain histocompatibility antigens are associated with poorer outcomes.
  • Nonsteroidal anti-inflammatory drugs NSAIDs
  • DMARDs Disease- modifying antirheumatic drugs
  • cytokines such as tumor necrosis factor-alpha (TNFa), interleukin-1 , interleukin-6 and granulocyte macrophage colony stimulating factor (GM-CSF), which lead to the activation and proliferation of immune cells, are found to be increased in the inflamed joint.
  • TNFa tumor necrosis factor-alpha
  • GM-CSF granulocyte macrophage colony stimulating factor
  • CSFs tumor necrosis factor alpha
  • RA tumor necrosis factor alpha
  • GM-CSF granulocyte-macrophage colony-stimulation factor
  • MOR103 is a fully human anti-GM-CSF antibody (Mol Immunol (2008) 46, 135-44; WO 2006/122797). MOR 103 is also in a clinical Phase lb trial for multiple sclerosis. The present invention describes the development of a clinically efficacious treatment regimen comprising MOR103 for RA. SUMMARY OF THE INVENTION
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention also provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM- CSF antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the anti-GM-CSF antibody is administered intravenously, optionally at a dosage of at least 1 .0 mg/kg, or at a dose of about 1 .0 mg/kg or about 1.5 mg/kg. In an embodiment, the anti-GM-CSD antibody is administered weekly, over at least four weeks.
  • the anti-GM-CSF antibody is administered subcutaneously, optionally at a dose of at least 2.0 mg/kg, or at a dose of about 2.0 mg/kg, about 3.0 mg/kg or about 4.0 mg/kg.
  • the anti-GM-CSF antibody is administered biweekly, monthly or bimonthly.
  • the antibody is administered at a fixed dose of about 75 mg, of about 100 mg, of about 150 mg, of about 200 mg, of about 300 mg or of about 400 mg. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • the dosage of anti-GM-CSF antibody administered to said patient and frequency of said administration is sufficient to provide and maintain a serum concentration of said antibody at at least 2 ⁇ g/ml in said patient over the duration of said treatment.
  • the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G ATYYAAS V KG (SEQ I D NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least
  • the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G ATYYAAS V KG (SEQ I D NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered intravenously at a dose of about 1 .0 mg/kg or at a dose of about 1.5 mg/kg and wherein said antibody in administered weekly over at least four weeks.
  • said anti-GM-CSF antibody is an antibody comprising an
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5mg/kg when administered weekly over at least four weeks, and wherein said anti-GM-CSF antibody is administered in combination with a DMARD, such as methotrexate.
  • a DMARD such as methotrexate
  • the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg.
  • the antibody of the present invention is administered intravenously at a dose of about 1 .0 mg/kg or a dose of about 1 .5 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient subcutaneously in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5mg/kg when administered weekly over at least four weeks, and wherein said anti-GM- CSF antibody is administered in combination with a DMARD, such as methotrexate.
  • a DMARD such as methotrexate
  • the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a dose of at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg.
  • the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
  • the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a fixed dose of about 40 mg, at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg.
  • Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • the present invention provides a method of treating a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM- CSF antibody subcutaneously at
  • the anti-GM-CSF antibody may be administered to said patient in a manner to achieve to a serum concentration of said antibody at at least 2 ⁇ g/ml in said patient over the duration of said treatment.
  • the antibody may be administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • Figure 1 shows the amino acid sequence and the DNA sequence of MOR04357.
  • Figure 2 shows the mean changes of the DAS28 score after four weeks (left panel) and after eight weeks (right panel) of treatment compared. DAS28 score changes are compared to baseline levels, i.e. disease status prior to treatment.
  • Figure 3 shows the average ACR20 score of all treatment arms after four weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
  • Figure 4 shows the average ACR20 score of all treatment arms after eight weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
  • GM-CSF and “GMCSF” refer to the protein known as GM-CSF or Granulocyte-macrophage colony-stimulating factor, having the following synonyms: Colony- stimulating factor 2, CSF2, GMCSF, GM-CSF, Granulocyte-macrophage colony-stimulating factor, MGCI 31935, MGC138897, Molgramostin, Sargramostim.
  • Human GM-CSF has the amino acid sequence of (UniProt P04141 ):
  • MOR103 is an anti-GM-CSF antibody whose amino acid sequence and DNA sequence is provided in Figure 1.
  • MOR103 and “MOR04357” and “MOR4357” are used as synonyms to describe the antibody shown in Figure 1 .
  • MOR04357 comprises an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • MOR04357 comprises a variable heavy chain of the sequence
  • the antibody used in the present invention is an antibody specific for GM-CSF. In other embodiments, the antibody used in the present invention is an antibody specific for a polypeptide encoding an amino acid sequence comprising SEQ ID NO.: 1.
  • binding affinity for antigen is of Kd value of 10 "9 mol/l or lower (e.g. 10 "10 mol/l), preferably with a Kd value of 10 "10 mol/l or lower (e.g. 10 "12 mol/l).
  • the binding affinity is determined with a standard binding assay, such as surface plasmon resonance technique (BIACORE ® ).
  • BIACORE ® surface plasmon resonance technique
  • the antibody used in the present invention is MOR103.
  • the antibody used in the present invention is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G AT YYAAS V KG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • the antibody used in the present invention is an antibody comprising a variable heavy chain of the sequence QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS
  • the antibody used in the present invention is an antibody which cross- competes with an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G ATYYAAS VKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G ATYYAAS VKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKY
  • the antibody used in the present invention is an antibody which binds to the same epitope like an antibody specific for GM-CSF comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G ATYYAAS V KG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • an antibody specific for GM-CSF comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence G I E N KYAG G ATYYAAS V KG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • Antibody fragments herein comprise a portion of an intact antibody which retains the ability to bind antigen.
  • Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they are uncontaminated by other immunoglobulins.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non- human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non- human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence, except for FR substitution(s) as noted above.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin.
  • a "human antibody” herein is one comprising an amino acid sequence structure that corresponds with the amino acid sequence structure of an antibody obtainable from a human B- cell, and includes antigen-binding fragments of human antibodies.
  • Such antibodies can be identified or made by a variety of techniques, including, but not limited to: production by transgenic animals (e.g., mice) that are capable, upon immunization, of producing human antibodies in the absence of endogenous immunoglobulin; selection from phage display libraries expressing human antibodies or human antibody; generation via in vitro activated B; and isolation from human antibody producing hybridomas.
  • the antibody used in the present invention is a monoclonal antibody.
  • the antibody used in the present invention is a chimeric, a humanized or a human antibody. In preferred embodiments, the antibody used in the present invention is a human antibody.
  • the antibody used in the present invention is administered in combination with an additional drug that treats RA.
  • the additional drug may be one or more medicaments, and include, for example, immunosuppressive agents, non-steroidal anti-inflammatory drugs (NSAIDs), disease modifying anti-rheumatic drugs (DMARDs) such as methotrexate (MTX), anti-B-cell surface marker antibodies, such as anti-CD20 antibodies (e.g. rituximab), TNF-alpha-inhibitors, corticosteroids, and co-stimulatory modifiers, or any combination thereof.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • DMARDs disease modifying anti-rheumatic drugs
  • MTX methotrexate
  • anti-B-cell surface marker antibodies such as anti-CD20 antibodies (e.g. rituximab)
  • TNF-alpha-inhibitors e.g. rituximab
  • corticosteroids e.g. rituximab
  • additional drugs are generally used in the same dosages and with administration routes as used hereinbefore and hereinafter. If such additional drugs are used at all, preferably, they are used in lower amounts than if the first medicament were not present, especially in subsequent dosings beyond the initial dosing with the first medicament, so as to eliminate or reduce side effects caused thereby.
  • the combined administration of an additional drug includes co-administration (concurrent administration), using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents (medicaments) simultaneously exert their biological activities.
  • DMARD refers to "Disease-Modifying Anti-Rheumatic Drugs" and includes among others hydroxycloroquine, sulfasalazine, methotrexate, leflunomide, azathioprine, D- penicillamine, gold salts (oral), gold salts (intramuscular), minocycline, cyclosporine including cyclosporine A and topical cyclosporine, and TNF-inhibitors, including salts, variants, and derivatives thereof.
  • Exemplary DMARDs herein are non-biological, i.e. classic, DMARDs, including, azathioprine, chloroquine, hydroxychloroquine, leflunomide, methotrexate and sulfasalazine.
  • Methotrexate is an especially preferred DMARD of the present invention. Therefore, in certain embodiments, the antibody used in the present invention is administered in combination with a DMARD. In other embodiments, the antibody used in the present invention is administered in combination with methotrexate.
  • TNF-inhibitor refers to an agent that inhibits, to some extent, a biological function of TNF-alpha, generally through binding to TNF-alpha and/or its receptor and neutralizing its activity.
  • TNF inhibitors include etanercept (ENBREL ® ), infliximab (REMICADE ® ), adalimumab (HUMIRA ® ), certolizumab pegol (CIMZIA ® ), and golimumab (SIMPONI ® ).
  • the antibody of the present invention may be administered in different suitable forms.
  • Potential forms of administration include systemic administration (subcutaneous, intravenous, intramuscular), oral administration, inhalation, transdermal administration, topical application (such as topical cream or ointment, etc.) or by other methods known in the art.
  • the doses (in mg/kg) specified in the present invention refer to milligrams of antibody per kilogram of body weight of the patient.
  • In vitro cell based assays showed that an anti-GM- CSF antibody (MOR103) is capable of inhibiting several GM-CSF mediated responses.
  • Evaluated responses include TF-1 cell proliferation, STAT5 phosphorylation, polymorphonuclear neutrophils (PMN) migration, PMN up-regulation of CD1 1 b, monocyte up-regulation of MHC II, and eosinophil survival.
  • PMN polymorphonuclear neutrophils
  • GM-CSF concentrations up to 1 ng/ml were applied in such studies.
  • GM-CSF levels in the synovial fluid of RA patients were reported to be ⁇ 500 pg/ml. It is reasonable to consider that similar GM-CSF concentrations as used in these in vitro studies are present in affected tissues of RA patients
  • a specific anti-GM-CSF antibody (MOR103) has been administered to patients with active rheumatoid arthritis who received 4 intravenous weekly doses of 0.3, 1 , and 1.5 mg/kg.
  • the anti-GM-CSF antibody showed significant clinical efficacy on DAS28, EULAR, ACR20, ACR50, ACR70 and tender joint counts following once a week dosing with 1 and 1 .5 mg/kg as compared to placebo.
  • the antibody of the present invention is administered intravenously. In other embodiments, the antibody of the present invention is administered subcutaneously.
  • a concentration that leads to a certain level of the antibody in the blood when administered intravenously corresponds to about 50- 76% of the blood concentration achieved when the same antibody concentration is administered subcutaneously (Meibohm, B.: Pharmacokinetics and Pharmacodynamics of Biotech Drugs, Wiley-VCH, 2006).
  • this ratio was determined to be 52%, i.e. a given concentration administered subcutaneously leads to a blood concentration which is equivalent to about 52% of the blood concentration when the same given concentration is administered intravenously. Therefore, the concentration of a subcutaneous formulation needs to be about twice as high to achieve the same drug blood level as compared to an intravenous formulation.
  • the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • said blood concentration to be achieved is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a doses of at least 0.4, 0.5, 0.6, 0.7, 0.8 or 0.9 mg/kg when administered weekly over at least four weeks.
  • the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least two weeks or at least three weeks.
  • the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered biweekly over at least two weeks or at least four weeks.
  • the antibody of the present invention is administered intravenously.
  • the antibody of the present invention is administered intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1 .0 mg/kg.
  • the antibody of the present invention is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg.
  • the antibody of the present invention is administered subcutaneously.
  • Various dosing regimen have been simulated using the subcutaneous delivery of MOR103 in order to produce plasma concentrations that are similar those obtained after 1 mg/kg iv, a dose that was efficacious in RA.
  • the majority of simulations produce trough concentration values greater than 2 ug/mL, a value that is believed to be the minimum blood concentration that is required to produce efficacy in the context of an anti- GM-CSF antibody.
  • the antibody of the present invention is administered subcutaneously at a dose at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg. In other embodiments, the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. In certain embodiments, the antibody of the present invention is subcutaneously administered biweekly, monthly or bimonthly.
  • the antibody of the present invention is administered subcutaneously at a fixed dose.
  • the antibody is administered at a certain, fixed, concentration, i.e. without taking into account a patient's body weight.
  • the antibody of the present invention is administered at a fixed dose of between 40 mg and 400 mg, optionally at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg.
  • Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week. Typically, the antibody will be administered weekly at a fixed dose.
  • the antibody will be administered weekly, at a fixed subcutaneous dose of 40, 56, 70, 75 100, 140, 150, 180, 200, 210, or 280 mg.
  • the antibody will be administered biweekly, at a fixed subcutaneous dose of 70, 75, 100, 140, 150, 180, 200, 210, 280 or 300 mg.
  • the antibody will be administered monthly, at a fixed subcutaneous dose of 100, 140, 150, 180, 200, 210, 280, 300, 320, 350. 360 or 400 mg.
  • the antibody is administered in a dose sufficient to maintain trough concentration of antibody of at least 2 ug/mL.
  • the trough concentration of antibody may be maintained at 2.0 ug/mL, 2.5 ug/mL, 3.0 ug/mL, 3.5 ug/mL, 4.0 ug/mL, 4.5 ug/mL or 5.0 ug/mL, during the course of therapy.
  • the antibody will be administered weekly, at a fixed subcutaneous dose of 28 or 35 mg,
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • drug and “medicament” refer to an active drug to treat rheumatoid arthritis or joint damage or symptoms or side effects associated with RA.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of the active ingredient or ingredients, i.e. the antibody of the present invention, to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile.
  • the antibody herein is preferably recombinantly produced in a host cell transformed with nucleic acid sequences encoding its heavy and light chains (e.g. where the host cell has been transformed by one or more vectors with the nucleic acid therein).
  • the preferred host cell is a mammalian cell, most preferably a PER.C6 cell.
  • Therapeutic formulations of the antibody of the present invention are prepared for storage by mixing the antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, histidine and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) poly
  • the present invention provides a pharmaceutical composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier and/or excipient for use in any of the methods provided in the present invention.
  • the formulation for the antibody of the present invention consists of 30 mM histidine, pH 6.0, 200 mM sorbitol and 0.02% Tween-80.
  • the formulation for the antibody of the present invention consists of PBS, pH 6.2 (0.2 g/l KCI, 0.96 g/l KH 2 P0 4 , 0.66 g/l Na 2 HP0 4 x 7H 2 0, 8 g/l NaCI).
  • Example 1 Design and concept of a clinical Phase lb/Phase 11 a trial
  • Primary outcome measures were the adverse event rate and the safety profile. Secondary outcome measures included DAS28 scores, ACR scores and EULAR28 response criteria.
  • the clinical trial comprised three treatment arms.
  • each treatment arm patient received either placebo or MOR103.
  • the MOR103 doses were 0.3 mg/kg body weight for treatment arm 1 , 1.0 mg/kg body weight for treatment arm 2 and 1.5 mg/kg body weight for treatment arm 3.
  • MOR103 and placebo were administered intravenously, weekly with 4 doses in total.
  • MOR103 experimental MOR103 0.3 mg/kg or placebo iv x 4 doses Biological: MOR103 0.3 mg/kg or placebo
  • MOR103 experimental MOR103 1.0 mg/kg or placebo iv x 4 doses Biological: MOR103 1.0 mg/kg or placebo
  • MOR103 experimental MOR103 1.5 mg/kg or placebo iv x 4 doses Biological: MOR103 1.5 mg/kg or placebo
  • Example 2 Patient recruitment and patient population
  • DAS28 score Prior to administration of MOR103 or the placebo the disease activity of all patients was measured according to accepted guidelines by calculating the DAS28 score, a 28-joint Disease Activity Score (see e.g. Ann Rheum Dis (2009) 68, 954-60). DAS28 score is a validated and commonly used tool to quantify the disease status of RA patients. The average DAS28 score was comparable for all treatment arms.
  • MOR103 Based on the available observed safety data, MOR103 showed a favorable safety profile among all doses tested. The key observations are as follows:
  • ACR criteria measure improvement in tender or swollen joint counts and improvement in certain other parameters.
  • the procedure to measure ACR scores is highly standardized. The present clinical trial applied the respective applicable guidelines. Results are depicted in Figures 3 and 4. A higher score corresponds to an improvement in the severity of the disease.
  • the ACR scores show a strong clinical improvement of patients' condition upon treatment with either 1 .0 mg/kg MOR103 or 1.5 mg/kg MOR103.
  • the improvement after 4 weeks is highly significant for the 1.0 mg/kg group (p ⁇ 0.0001 ).
  • the ACR20 scores confirm the surprising finding that the efficacy of MOR103 can already be shown with a comparably low number or patients in each treatment arm and a comparably short treatment period.
  • MOR103 is administered to patients intravenously at a dose of 0.5 mg/kg (treatment arm 1 ) and 0.75 mg/kg (treatment arm 2). All other parameters are identical to Example 1 .
  • Both treatment arms show a favorable safety profile and demonstrate clinical efficacy as measured by DAS28 scores and ACR20 scores.
  • Example 7 Clinical trial with a sub-cutaneous formulation of MOR103
  • MOR103 is administered to patients at 1.5 mg/kg, 2.0 mg/kg, 3.0 mg/kg and 4.0 mg/kg.
  • the drug is administered sub-cutaneously, either biweekly, monthly or bimonthly. All other parameters are identical to Example 1. All treatment arms show a favorable safety profile and demonstrate clinical efficacy as measured by DAS28 scores and ACR20 scores.
  • Example 8 Clinical trial with a sub-cutaneous formulation of MOR103 at a fixed dose
  • Example 7 is repeated with a fixed dose of MOR103.
  • MOR103 is administered to patients at fixed dose of 75 mg, of 100 mg, of 150 mg, of 200 mg, of 300 mg and of 400 mg.
  • the drug is administered sub-cutaneously every week, every second week, every fourth week or every sixth week. All other parameters are identical to the Examples described herein above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Endocrinology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
PCT/EP2013/069501 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis WO2014044768A1 (en)

Priority Applications (27)

Application Number Priority Date Filing Date Title
BR112015006189-3A BR112015006189B1 (pt) 2012-09-20 2013-09-19 Uso de um anticorpo anti-gm-csf
MX2015003644A MX2015003644A (es) 2012-09-20 2013-09-19 Tratamiento para artritis reumatoide.
KR1020217005022A KR20210021153A (ko) 2012-09-20 2013-09-19 류마티스 관절염에 대한 치료
CA2884124A CA2884124A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
KR1020157009911A KR20150056846A (ko) 2012-09-20 2013-09-19 류마티스 관절염에 대한 치료
MA37946A MA37946B1 (fr) 2012-09-20 2013-09-19 Utilisation d’un anticorps anti-gm-csf pour le traitement de la polyarthrite rhumatoïde
KR1020237008704A KR20230041086A (ko) 2012-09-20 2013-09-19 류마티스 관절염에 대한 치료
IN657KON2015 IN2015KN00657A (uk) 2012-09-20 2013-09-19
KR1020227006147A KR20220028177A (ko) 2012-09-20 2013-09-19 류마티스 관절염에 대한 치료
CN201380049042.8A CN104995210A (zh) 2012-09-20 2013-09-19 类风湿关节炎的治疗
EP18155371.0A EP3345923A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis with anti-gm-csf antibody
EP21169696.8A EP3916013A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis with anti-gm-csf antibody
EA201590359A EA031489B1 (ru) 2012-09-20 2013-09-19 Способ лечения ревматоидного артрита
AU2013320261A AU2013320261A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
JP2015532412A JP2015533806A (ja) 2012-09-20 2013-09-19 関節リウマチの治療
UAA201502131A UA117228C2 (uk) 2012-09-20 2013-09-19 Фармацевтична композиція, що містить антитіло до gm-csf
SG11201501595YA SG11201501595YA (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
EP13763100.8A EP2897977A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
US14/429,996 US20150246969A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
IL237554A IL237554B (en) 2012-09-20 2015-03-04 A pharmaceutical preparation containing an antibody against gm-CSF for the treatment of a patient suffering from rheumatoid arthritis
ZA2015/01559A ZA201501559B (en) 2012-09-20 2015-03-06 Treatment for rheumatoid arthritis
PH12015500591A PH12015500591B1 (en) 2012-09-20 2015-03-18 Treatment for rheumatoid arthritis
CR20150153A CR20150153A (es) 2012-09-20 2015-03-20 Tratamiento para artitris reumatoide
HK15108769.5A HK1208231A1 (en) 2012-09-20 2015-09-09 Treatment for rheumatoid arthritis
AU2016250388A AU2016250388B2 (en) 2012-09-20 2016-10-26 Treatment for rheumatoid arthritis
US15/880,726 US10913792B2 (en) 2012-09-20 2018-01-26 Treatment for rheumatoid arthritis
US17/144,685 US20210130451A1 (en) 2012-09-20 2021-01-08 Treatment for rheumatoid arthritis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP12185235 2012-09-20
EP12185235.4 2012-09-20
US201261703871P 2012-09-21 2012-09-21
US61/703,871 2012-09-21

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/429,996 A-371-Of-International US20150246969A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
US15/880,726 Continuation US10913792B2 (en) 2012-09-20 2018-01-26 Treatment for rheumatoid arthritis

Publications (1)

Publication Number Publication Date
WO2014044768A1 true WO2014044768A1 (en) 2014-03-27

Family

ID=46963526

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/069501 WO2014044768A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis

Country Status (23)

Country Link
US (3) US20150246969A1 (uk)
EP (3) EP2897977A1 (uk)
JP (3) JP2015533806A (uk)
KR (3) KR20220028177A (uk)
CN (2) CN104995210A (uk)
AU (2) AU2013320261A1 (uk)
BR (1) BR112015006189B1 (uk)
CA (1) CA2884124A1 (uk)
CL (2) CL2015000696A1 (uk)
CR (1) CR20150153A (uk)
DO (1) DOP2015000070A (uk)
EA (1) EA031489B1 (uk)
HK (1) HK1208231A1 (uk)
IL (1) IL237554B (uk)
IN (1) IN2015KN00657A (uk)
MA (1) MA37946B1 (uk)
MX (2) MX2015003644A (uk)
MY (1) MY175388A (uk)
PE (1) PE20151079A1 (uk)
PH (1) PH12015500591B1 (uk)
SG (2) SG10201803778PA (uk)
UA (1) UA117228C2 (uk)
WO (1) WO2014044768A1 (uk)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014068026A1 (en) 2012-10-31 2014-05-08 Amgen Research (Munich) Gmbh Liquid formulation comprising gm-csf neutralizing compound
WO2015028657A1 (en) * 2013-08-30 2015-03-05 Takeda Gmbh Antibodies neutralizing gm-csf for use in the treatment of rheumatoid arthritis or as analgesics
US20150259397A1 (en) * 2014-03-11 2015-09-17 Theraly Pharmaceuticals Inc. Long acting trial receptor agonists for treatment of autoimmune diseases
CN106659785A (zh) * 2014-05-07 2017-05-10 武田有限公司 包含gm‑csf中和化合物的液体制剂
WO2017076804A1 (en) * 2015-11-02 2017-05-11 Glaxosmithkline Intellectual Property Development Limited Treatment paradigm
US9833410B2 (en) 2012-10-31 2017-12-05 Takeda Gmbh Lyophilized formulation comprising GM-CSF neutralizing compound
WO2018050111A1 (en) * 2016-09-19 2018-03-22 I-Mab Anti-gm-csf antibodies and uses thereof
US11007251B2 (en) 2015-12-17 2021-05-18 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
US11084879B2 (en) 2016-04-07 2021-08-10 The Johns Hopkins University Compositions and methods for treating pancreatitis and pain with death receptor agonists
WO2021204649A1 (en) 2020-04-06 2021-10-14 Glaxosmithkline Intellectual Property Development Limited Gm-csf antagonists for use in the treatment of severe pulmonary covid-19, cytokine release syndrome and/or acute respiratory distress syndrome

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IN2015KN00657A (uk) * 2012-09-20 2015-07-17 Morphosys Ag
EP3623382A1 (en) 2018-09-14 2020-03-18 Universität Zürich Ligands to gm-csf or gm-csf-receptor for use in leukemia in a patient having undergone allo-hct
US11655293B2 (en) * 2018-02-22 2023-05-23 Universitat Zurich Ligands to GM-CSF or GM-CSF-receptor for use in leukemia in a patient having undergone allo-HCT

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006122797A2 (en) 2005-05-18 2006-11-23 Morphosys Ag Anti-gm-csf antibodies and uses therefor
WO2008064321A2 (en) * 2006-11-21 2008-05-29 Kalobios Pharmaceuticals, Inc. Methods of treating chronic inflammatory diseases using a gm-csf antagonist
WO2009038760A2 (en) * 2007-09-18 2009-03-26 Amgen Inc. Human gm-csf antigen binding proteins
WO2010128035A1 (en) * 2009-05-05 2010-11-11 Morphosys Ag Treatment for multiple sclerosis

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
CN1142940C (zh) 1996-10-04 2004-03-24 安姆根有限公司 含有mpl配体的药物组合物
US7455836B2 (en) 2000-05-08 2008-11-25 The University Of Melbourne Method of treatment and agents useful for same
EP1916997B1 (en) 2005-08-05 2018-04-18 Amgen Inc. Stable aqueous protein pharmaceutical formulations and their preparation
EP2010214A4 (en) * 2006-04-10 2010-06-16 Abbott Biotech Ltd USES AND COMPOSITIONS FOR THE TREATMENT OF RHEUMATOID ARTHRITIS
US8093360B2 (en) * 2006-09-28 2012-01-10 Elusys Therapeutics, Inc. Antibodies that bind B. anthracis exotoxin, formulations thereof, and methods of use
AU2008232902B2 (en) * 2007-03-30 2013-10-03 Medlmmune, Llc Antibody formulation
EP2137325A1 (en) * 2007-04-02 2009-12-30 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
AU2008235044B2 (en) 2007-04-05 2011-08-25 Sandoz Ag Stable aqueous G-CSF formulations
US20090004189A1 (en) * 2007-06-18 2009-01-01 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
BRPI0820530A2 (pt) 2007-11-13 2015-06-16 Boehringer Ingelheim Int Anticorpos monoclonais que se ligam ao hgm-csf e composições medicinais compreendendo os mesmos
EP2196476A1 (en) * 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
BRPI0918103A2 (pt) * 2008-12-22 2016-07-05 Univ Melbourne "método para o tratamento da dor em um indivíduo"
CA2746827C (en) 2008-12-22 2018-01-23 The University Of Melbourne Osteoarthritis treatment
JP2010241718A (ja) 2009-04-03 2010-10-28 Kyowa Hakko Kirin Co Ltd 安定な抗体の水溶液製剤
SI2341061T1 (sl) 2009-12-31 2013-12-31 Arven Ilac Sanayi Ve Ticaret A.S. Nov postopek za pripravo G-CSF-ja (granulocitne kolonije stimulirajočega faktorja)
WO2011109365A2 (en) * 2010-03-01 2011-09-09 Progenics Pharmaceuticals, Inc. Concentrated protein formulations and uses thereof
BR112013033944A2 (pt) 2011-07-06 2017-12-19 Morphosys Ag combinação sinérgica
IN2015KN00657A (uk) * 2012-09-20 2015-07-17 Morphosys Ag

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006122797A2 (en) 2005-05-18 2006-11-23 Morphosys Ag Anti-gm-csf antibodies and uses therefor
WO2008064321A2 (en) * 2006-11-21 2008-05-29 Kalobios Pharmaceuticals, Inc. Methods of treating chronic inflammatory diseases using a gm-csf antagonist
WO2009038760A2 (en) * 2007-09-18 2009-03-26 Amgen Inc. Human gm-csf antigen binding proteins
WO2010128035A1 (en) * 2009-05-05 2010-11-11 Morphosys Ag Treatment for multiple sclerosis

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANN RHEUM DIS, vol. 68, 2009, pages 954 - 60
MEIBOHM, B.: "Pharmacokinetics and Pharmacodynamics of Biotech Drugs", 2006, WILEY-VCH
MOL IMMUNOL, vol. 46, 2008, pages 135 - 44
NAT REV IMMUNOL, vol. 8, 2008, pages 533 - 44
NAT REV RHEUMATOL, vol. 5, 2009, pages 554 - 9

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9833410B2 (en) 2012-10-31 2017-12-05 Takeda Gmbh Lyophilized formulation comprising GM-CSF neutralizing compound
US10758621B2 (en) 2012-10-31 2020-09-01 Amgen Research (Munich) Gmbh Liquid formulation comprising GM-CSF neutralizing compound
WO2014068026A1 (en) 2012-10-31 2014-05-08 Amgen Research (Munich) Gmbh Liquid formulation comprising gm-csf neutralizing compound
EP2914289B1 (en) * 2012-10-31 2019-05-22 Takeda GmbH Lyophilized formulation comprising gm-csf neutralizing compound
US9919051B2 (en) 2012-10-31 2018-03-20 Amgen Research (Munich) Gmbh Liquid formulation comprising GM-CSF neutralizing compound
US10745475B2 (en) 2013-08-30 2020-08-18 Takeda Gmbh Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
EP3889178A1 (en) * 2013-08-30 2021-10-06 Takeda GmbH Antibodies neutralizing gm-csf for use in the treatment of rheumatoid arthritis or as analgesics
US11795216B2 (en) 2013-08-30 2023-10-24 Takeda Pharmaceutical Company Limited Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
WO2015028657A1 (en) * 2013-08-30 2015-03-05 Takeda Gmbh Antibodies neutralizing gm-csf for use in the treatment of rheumatoid arthritis or as analgesics
US11299528B2 (en) * 2014-03-11 2022-04-12 D&D Pharmatech Inc. Long acting TRAIL receptor agonists for treatment of autoimmune diseases
US20150259397A1 (en) * 2014-03-11 2015-09-17 Theraly Pharmaceuticals Inc. Long acting trial receptor agonists for treatment of autoimmune diseases
EP3139960B1 (en) * 2014-05-07 2024-01-17 Takeda Pharmaceutical Company Limited Liquid formulation comprising gm-csf neutralizing compound
US11173208B2 (en) 2014-05-07 2021-11-16 Takeda Gmbh Liquid formulation comprising GM-CSF neutralizing compound
CN106659785A (zh) * 2014-05-07 2017-05-10 武田有限公司 包含gm‑csf中和化合物的液体制剂
JP2017514868A (ja) * 2014-05-07 2017-06-08 タケダ・ゲー・エム・ベー・ハーTakeda GmbH Gm−csf中和化合物を含む液体製剤
JP2018533588A (ja) * 2015-11-02 2018-11-15 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited 治療パラダイム
WO2017076804A1 (en) * 2015-11-02 2017-05-11 Glaxosmithkline Intellectual Property Development Limited Treatment paradigm
US11007251B2 (en) 2015-12-17 2021-05-18 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
US11084879B2 (en) 2016-04-07 2021-08-10 The Johns Hopkins University Compositions and methods for treating pancreatitis and pain with death receptor agonists
US10889641B2 (en) 2016-09-19 2021-01-12 I-Mab Biopharma Co., Ltd. Anti-GM-CSF antibodies and uses thereof
US10647767B2 (en) 2016-09-19 2020-05-12 I-Mab Biopharma Co., Ltd. Anti-GM-CSF antibodies and uses thereof
WO2018050111A1 (en) * 2016-09-19 2018-03-22 I-Mab Anti-gm-csf antibodies and uses thereof
US11926662B2 (en) 2016-09-19 2024-03-12 I-Mab Biopharma (Hangzhou) Co., Ltd. Anti-GM-CSF antibodies and uses thereof
WO2021204649A1 (en) 2020-04-06 2021-10-14 Glaxosmithkline Intellectual Property Development Limited Gm-csf antagonists for use in the treatment of severe pulmonary covid-19, cytokine release syndrome and/or acute respiratory distress syndrome

Also Published As

Publication number Publication date
UA117228C2 (uk) 2018-07-10
HK1208231A1 (en) 2016-02-26
US20180230208A1 (en) 2018-08-16
JP2018138551A (ja) 2018-09-06
AU2016250388B2 (en) 2018-07-19
MA37946B1 (fr) 2018-09-28
DOP2015000070A (es) 2015-08-16
KR20150056846A (ko) 2015-05-27
IN2015KN00657A (uk) 2015-07-17
EA201590359A1 (ru) 2015-08-31
BR112015006189A2 (pt) 2019-11-26
EA031489B1 (ru) 2019-01-31
PH12015500591A1 (en) 2015-05-11
US20210130451A1 (en) 2021-05-06
JP2015533806A (ja) 2015-11-26
CA2884124A1 (en) 2014-03-27
KR20230041086A (ko) 2023-03-23
AU2016250388A1 (en) 2016-11-17
PH12015500591B1 (en) 2015-05-11
MY175388A (en) 2020-06-23
SG11201501595YA (en) 2015-05-28
KR20220028177A (ko) 2022-03-08
MX2015003644A (es) 2015-09-25
CN104995210A (zh) 2015-10-21
AU2013320261A1 (en) 2015-04-09
EP3916013A1 (en) 2021-12-01
PE20151079A1 (es) 2015-08-07
JP2022169709A (ja) 2022-11-09
CL2018000933A1 (es) 2018-07-20
US10913792B2 (en) 2021-02-09
EP3345923A1 (en) 2018-07-11
US20150246969A1 (en) 2015-09-03
CN109999195A (zh) 2019-07-12
EP2897977A1 (en) 2015-07-29
MX2021006111A (es) 2021-07-07
IL237554A0 (en) 2015-04-30
CR20150153A (es) 2015-09-14
IL237554B (en) 2019-12-31
BR112015006189B1 (pt) 2022-04-05
SG10201803778PA (en) 2018-06-28
MA37946A1 (fr) 2018-01-31
CL2015000696A1 (es) 2015-10-23

Similar Documents

Publication Publication Date Title
US20210130451A1 (en) Treatment for rheumatoid arthritis
US20200231666A1 (en) Treatment paradigm
JP6554155B2 (ja) Il−17アンタゴニストを使用して汎発性膿疱性乾癬(gpp)を処置する方法
JP2024001125A (ja) Il-17アンタゴニストを用いて初発プラーク型乾癬を治療する方法
JP7132256B2 (ja) 関節リウマチの治療
TW202114734A (zh) Il-17拮抗劑治療自身免疫疾病的方法
NZ744721A (en) Treatment for rheumatoid arthritis
NZ744721B2 (en) Treatment for rheumatoid arthritis
WO2021216374A1 (en) Pan-elr+ cxc chemokine antibodies for the treatment of respiratory disease
JP2024516019A (ja) 抗baffr抗体を使用する全身性エリテマトーデスのための治療
TW202302147A (zh) 使用抗baffr抗體治療狼瘡腎炎

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13763100

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2884124

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 237554

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 201590359

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 000350-2015

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 2015532412

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12015500591

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2015000696

Country of ref document: CL

Ref document number: 15063364

Country of ref document: CO

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14429996

Country of ref document: US

Ref document number: 37946

Country of ref document: MA

Ref document number: MX/A/2015/003644

Country of ref document: MX

Ref document number: CR2015-000153

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 2013763100

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015006189

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: A201502131

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 2013320261

Country of ref document: AU

Date of ref document: 20130919

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: IDP00201502214

Country of ref document: ID

ENP Entry into the national phase

Ref document number: 20157009911

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112015006189

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150319

ENPC Correction to former announcement of entry into national phase, pct application did not enter into the national phase

Ref document number: 112015006189

Country of ref document: BR

Kind code of ref document: A2

Free format text: ANULADA A PUBLICACAO CODIGO 1.3 NA RPI NO 2426 DE 04/07/2017 POR TER SIDO INDEVIDA.

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112015006189

Country of ref document: BR

Kind code of ref document: A2

Free format text: APRESENTAR, EM ATE 60 (SESSENTA) DIAS, DOCUMENTOS COMPROBATORIOS QUE EXPLIQUEM E REGULARIZEM A DIVERGENCIA NO NOME DO INVENTOR CONSTANTE NA PUBLICACAO INTERNACIONAL WO/2014/044768 DE 27/03/2014 COMO STEFAN HAERTLE E O CONSTANTE NO FORMULARIO DA PETICAO INICIAL NO 860150045684 DE 19/03/2015 COMO STEFAN HAERTLE UMA VEZ QUE NAO HOUVE ENVIO DE DOCUMENTO COMPROVANDO QUE OS NOMES CORRETOS DO INVENTOR E O DECLARADO NA ENTRADA NACIONAL.

ENP Entry into the national phase

Ref document number: 112015006189

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150319